Proteomics reveals a therapeutic vulnerability via the combined blockade of APE1 and autophagy in lung cancer A549 cells

11 16 0
Proteomics reveals a therapeutic vulnerability via the combined blockade of APE1 and autophagy in lung cancer A549 cells

Đang tải... (xem toàn văn)

Tài liệu hạn chế xem trước, để xem đầy đủ mời bạn chọn Tải xuống

Thông tin tài liệu

Drug resistance is a major cause of therapeutic failure that is often associated with elevated autophagy and apurinic/apyrimidinic endonuclease 1 (APE1) expression. Herein, we investigated the role of APE1 and autophagy in A549 cells treated with cisplatin.

Pan et al BMC Cancer (2020) 20:634 https://doi.org/10.1186/s12885-020-07111-w RESEARCH ARTICLE Open Access Proteomics reveals a therapeutic vulnerability via the combined blockade of APE1 and autophagy in lung cancer A549 cells Shu-Ting Pan1†, Ji Zhou2†, Fang Yang2, Shu-Feng Zhou3* and Tao Ren4* Abstract Background: Drug resistance is a major cause of therapeutic failure that is often associated with elevated autophagy and apurinic/apyrimidinic endonuclease (APE1) expression Herein, we investigated the role of APE1 and autophagy in A549 cells treated with cisplatin Methods: SILAC proteomics was applied to obtain a panoramic view of cisplatin treatment in KRASG12S-mutant A549 cells Quantity analysis of cellular apoptosis and autophagy was based on flow cytometry Western blotting was used to examine the expression levels of apoptosis- and autophagy-related proteins, as well as those of APE1 Knockdown of APE1 was achieved by RNA interference Immunoprecipitation was further employed to reveal the molecular interaction of APE1, p53, and LC3 when A549 cells were exposed to cisplatin Results: SILAC proteomics revealed that 72 canonical pathways, including base excision repair (BER) and autophagy signalling pathways, were regulated after cisplatin treatment in A549 cells Cisplatin markedly induced autophagy and apoptosis in A549 cells, accompanied by remarkable APE1 increase Suppression of autophagy enhanced the inhibition effect of cisplatin on cell growth, proliferation, and colony formation; however, APE1 inhibition enhanced the expression of LC3-I/II, suggesting that APE1 and autophagy are compensatory for cell survival to evade the anticancer action of cisplatin Immunoprecipitation results revealed the triple complex of APE1-p53-LC3 in response to cisplatin plus CQ in A549 cells Dual inhibition of APE1 and autophagy significantly enhanced cisplatin-induced apoptosis, which eventually overcame drug resistance in cisplatin-resistant A549 cells Conclusions: Dual inhibition of APE1 and autophagy greatly enhances apoptosis in parental KRASG12S-mutant A549 cells and cisplatin-resistant A549 cells via regulation of APE1-p53-LC3 complex assembly, providing therapeutic vulnerability to overcome cisplatin resistance in the context of KRASG12S-mutant lung cancer Keywords: Cisplatin, APE1, Autophagy, Chemotherapy, Non-small cell lung cancer, Apoptosis * Correspondence: szhou@health.usf.edu; szhou@hqu.edu.cn; rentao509@cmc.edu.cn † Shu-Ting Pan and Ji Zhou contributed equally to this work Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, 12901 Bruce B Downs Boulevard, Tampa, Florida 33612, USA Oncology Department, The First Affiliated Hospital, Chengdu Medical College, 278 Baoguang St, Xindu Distr, Chengdu 610500, Sichuan, China Full list of author information is available at the end of the article © The Author(s) 2020 Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/ The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data Pan et al BMC Cancer (2020) 20:634 Background Lung cancer is the leading cause of cancer-related death and remains a major clinical challenge with increasing incidence and mortality [1, 2] Due to drug resistance, recurrence, and metastasis, the treatment efficacy of lung cancer remains unsatisfactory A better understanding of the aetiology, pathogenesis, and molecular targets is required to develop novel therapeutic modalities Somatic gene mutations, including KRAS, EGFR, and TP53 mutations, is a major driver of lung cancer initiation [3] Accumulating evidence has shown that not all gene mutations occur equally In particular, compelling evidence suggests that RAS mutants function in an allele-specific manner, justifying the acquirement of a RAS allele-specific approach for RAS-driven cancer therapy [4–6] Given the feature of allele specificity and the pivotal role of RAS in cellular events, including cell growth, cell survival, cell senescence, and cell death, novel strategies in a RAS allele-dependent manner are still required Autophagy is a cell survival-promoting mechanism following harsh stimuli and has been deeply implicated in cancer development and therapy [7–9] Recently, targeting autophagy has been in the spotlight for cancer therapy via pharmacological inhibition alone or combination with other therapeutics [10, 11], providing insight into lung cancer therapy development Cisplatin is one of the most frequently administered chemotherapeutic drugs for many solid tumours, including lung cancer Mechanically, cisplatin kills cancer cells via interference with DNA synthesis and repair, subsequently inducing cell apoptosis [12] However, there is limited clinical efficacy for cisplatinbased therapy because of drug resistance [13] Several key factors contribute to cisplatin resistance, including autophagy [14] and apurinic/apyrimidinic endonuclease (APE1) [15] APE1 is a multifunctional protein with two major activities, DNA repair and transcriptional regulation [16] Importantly, APE1 is often overexpressed in many tumours, contributing to disease progression, chemoresistance and a poor prognosis [15, 17–20] Our previous study found that APE1 is highly expressed in non-small cell lung cancer (NSCLC) Moreover, APE1 is a prognostic risk factor indicated by a poor overall survival [15, 19] Herein, targeting APE1 might represent a therapeutic vulnerability for lung cancer, particularly, cisplatin-resistant lung cancer Thus, based on the aforementioned details, we hypothesized that APE1 and autophagy may contribute to lung cancer progression and drug resistance and that combined blockade of APE1 and autophagy enhances the therapeutic effect of cisplatin and overcomes cisplatin resistance in lung cancer In the present study, we applied quantitative proteomics to identify the proteomic responses to cisplatin treatment in KRASG12S-mutant A549 cells Both APE1 and autophagy were involved in Page of 11 the cellular responses to cisplatin exposure In A549 cells and cisplatin-resistant A549 cells, cisplatin-induced apoptosis was significantly enhanced via the combination of autophagy inhibition by chloroquine (CQ) and APE1 knockdown by siRNA with the involvement of p53 activation Methods Chemicals and reagents CDDP was purchased from Selleckchem Inc (Houston, TX, USA) 13C6-L-lysine, L-lysine, 13C615N4-L-arginine, Larginine, Dulbecco’s modified Eagle’s medium (DMEM)/ F12 for SILAC, APE1 siRNA, dimethyl sulfoxide (DMSO), 2-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), bovine serum albumin, and Dulbecco’s phosphate-buffered saline (PBS) were obtained from Sigma-Aldrich (St Louis, MO, USA) 6-Diamidino-2-phenylindole (DAPI), Opti-minimal Essential Medium (MEM), Lipofectamine 2000, and the negative control siRNA were purchased from Invitrogen Inc (Carlsbad, CA, USA) The Annexin V-phycoerythrin (PE) apoptosis detection kit was purchased from BD Biosciences Inc (San Jose, CA, USA) The Cyto-ID® Autophagy detection kit was obtained from Enzo Life Sciences Inc (Farmingdale, NY, USA) The Western blotting substrate, Pierce™ bicinchoninic acid (BCA) protein assay kit, skim milk, and radioimmunoprecipitation assay buffer (RIPA) were purchased from Thermo Fisher Scientific Inc (Hudson, NH, USA) The polyvinylidene difluoride (PVDF) membrane was obtained from Bio-Rad Inc (Hercules, CA, USA) The antibody against human β-actin was obtained from Santa Cruz Biotechnology Inc (Dallas, TX, USA) The remaining primary antibodies for signalling proteins related to apoptosis and autophagy were purchased from Cell Signaling Technology Inc (Beverly, MA, USA) Cell line and cell culture The human lung cancer cell line A549 (KRASG12S) was obtained from Chinese Academy of Science Cellbank (Shanghai, China) and was cultured in RPMI1640 medium supplemented with 10% heat-inactivated foetal bovine serum (FBS) The cells were maintained at 37 °C in a 5% CO2/95% air humidified incubator Cell viability determination The MTT assay was used to evaluate cell viability Briefly, cells were seeded in 96-well plates at a density of 7.0 × 103 cells/well After 24 h of incubation, the cells were treated for 48 h The absorbance was measured using a Synergy™H4 Hybrid microplate reader (BioTek, Winooski, VT, USA) at wavelengths of 560 nm (MTT formazan) and 670 nm (background) Pan et al BMC Cancer (2020) 20:634 Page of 11 Quantitative proteomics Western blotting assay Quantitative proteomic experiments were performed using a stable isotope labelling by amino acids in cell culture (SILAC)-based approach to identify the molecular targets of CDDP in the treatment of A549 cells as previously described [21] Briefly, A549 cells were cultured in DMEM/F12 medium (for SILAC) with (heavy) or without (light) stable isotope-labelled amino acids (13C6 L-lysine and 13C615N4 L-arginine) and 10% dialyzed FBS After treatment with CDDP (5 μM) for 24 h., the cell samples were harvested, lysed, and quantified Next, an equal amount of heavy and light protein samples were combined to reach a total volume of 50 μL containing 400 μg of protein, and the combined protein sample was digested and desalted Next, the peptide mixtures (5 μL) were subjected to the hybrid linear ion trap The peptide SILAC ratio was calculated using MaxQuant version 1.2.0.13 The proteins were identified using Scaffold 4.3.2, and the pathway was analysed using ingenuity pathway analysis (IPA) from QIAGEN Inc The protein expression level was examined using Western blotting Protein samples were extracted using RIPA buffer, the protein concentrations were measured using the BCA kit, and an equal amount of protein was separated by SDS-PAGE The corresponding primary and secondary antibodies were applied to evaluate the expression levels of targeted proteins Visualization was performed using the Bio-Rad ChemiDoc™ XRS system, and the blot bands were analysed using Image Lab 3.0 RNA interference Small interfering RNA-mediated gene silencing was performed to investigate the role of APE1 in cisplatininduced apoptosis and autophagy in A549 cells according to the manufacturer’s instructions A549 cells were transfected with the negative control siRNA and APE1siRNA using Lipofectamine 2000 The protein samples were collected and kept at − 80 °C for further analysis Immunoprecipitation Quantification of cellular apoptosis Cell apoptosis was evaluated using the Annexin V-PE apoptosis detection kit as previously described [21] Briefly, the cells were collected after treatment and resuspended in 1× binding buffer with μL of Annexin VPE and μL of 7-amino-actinomycin D (7-AAD) at × 105 cells/mL in a total volume of 150 μL The cells were gently mixed and incubated in the dark for 15 at room temperature The binding buffer (100 μL) was then added to each tube, and the number of apoptotic cells was quantified using flow cytometry and collecting 10, 000 events for analysis Quantification of cellular autophagy Cell autophagy was examined using flow cytometry as previously described [21] Briefly, the cells were collected after treatment and resuspended in 250 μL of assay buffer containing 5% FBS, and Cyto-ID® Green stain solution (250 μL) was added to each tube and mixed gently After 20 of incubation at room temperature in the dark, the cells were collected by centrifugation, washed once and analysed using the green (FL1) channel of flow cytometry The interaction between APE1 and p53 was examined using immunoprecipitation as previously described [22] After 24 h of treatment, A549 cells were lysed in pre-chilled cell lysis buffer [50 mM Tris-HCl (pH 7.4), 150 mM NaCl, mM EDTA, 1% NP40, protease inhibitors] for The lysates were precleared with 20 μL of Proteins A/G (Invitrogen; Thermo Fisher Scientific, Inc.) at °C for 45 min, followed by incubation with APE1 or p53 antibody overnight at °C Following immunoprecipitation, the samples were incubated with protein G for h at °C Thereafter, the samples were washed with lysis buffer five times to remove any unprecipitated proteins before boiling in SDS buffer for The elution was analysed for precipitated APE1 or p53 protein using Western blotting analysis Normal rabbit IgG antibody was used as a negative control The antibodies used were as follows: APE1 (1:500), p53 (1:500), and normal rabbit IgG (1:1000) Statistical analysis The data were expressed as means ± standard deviation (SD) One-way analysis of variance (ANOVA) followed by Tukey’s multiple comparison procedure was used for comparisons of multiple groups The value of PMEK >ERK inhibition suggests a treatment strategy for RAS-driven cancers Nat Med 2019;25(4):620–7 11 Bryant KL, Stalnecker CA, Zeitouni D, Klomp JE, Peng S, Tikunov AP, Gunda V, Pierobon M, Waters AM, George SD, et al Combination of ERK and autophagy inhibition as a treatment approach for pancreatic cancer Nat Med 2019;25(4):628–40 12 Dasari S, Tchounwou PB Cisplatin in cancer therapy: molecular mechanisms of action Eur J Pharmacol 2014;740:364–78 Pan et al BMC Cancer (2020) 20:634 13 Hill DP, Harper A, Malcolm J, McAndrews MS, Mockus SM, Patterson SE, Reynolds T, Baker EJ, Bult CJ, Chesler EJ, et al Cisplatin-resistant triplenegative breast cancer subtypes: multiple mechanisms of resistance BMC Cancer 2019;19(1):1039 https://doi.org/10.1186/s12885-019-6278-9 14 Wang J, Wu GS Role of autophagy in cisplatin resistance in ovarian cancer cells J Biol Chem 2014;289(24):17163–73 15 Wang D, Xiang DB, Yang XQ, Chen LS, Li MX, Zhong ZY, Zhang YS APE1 overexpression is associated with cisplatin resistance in non-small cell lung cancer and targeted inhibition of APE1 enhances the activity of cisplatin in A549 cells Lung Cancer 2009;66(3):298–304 16 Li M, Wilson DM Human apurinic/apyrimidinic endonuclease Antioxid Redox Signal 2014;20(4):678–707 17 Wen X, Lu R, Xie S, Zheng H, Wang H, Wang Y, Sun J, Gao X, Guo L APE1 overexpression promotes the progression of ovarian cancer and serves as a potential therapeutic target Cancer Biomark 2016;17(3):313–22 18 Xiao X, Yang Y, Ren Y, Zou D, Zhang K, Wu Y rs1760944 Polymorphism in the APE1 Region is Associated with Risk and Prognosis of Osteosarcoma in the Chinese Han Population Sci Rep 2017;7(1):9331 https://doi.org/10.1038/ s41598-017-09750-9 19 Peng Y, Li Z, Zhang S, Xiong Y, Cun Y, Qian C, Li M, Ren T, Xia L, Cheng Y, et al Association of DNA base excision repair genes (OGG1, APE1 and XRCC1) polymorphisms with outcome to platinum-based chemotherapy in advanced nonsmall-cell lung cancer patients Int J Cancer 2014;135(11): 2687–96 20 Juhnke M, Heumann A, Chirico V, Hoflmayer D, Menz A, Hinsch A, HubeMagg C, Kluth M, Lang DS, Moller-Koop C, et al Apurinic/apyrimidinic endonuclease (APE1/Ref-1) overexpression is an independent prognostic marker in prostate cancer without TMPRSS2:ERG fusion Mol Carcinog 2017; 56(9):2135–45 21 He SJ, Shu LP, Zhou ZW, Yang T, Duan W, Zhang X, He ZX, Zhou SF Inhibition of Aurora kinases induces apoptosis and autophagy via AURKB/p70S6K/RPL15 axis in human leukemia cells Cancer Lett 2016;382(2):215–30 22 Zhai D, Cui C, Xie L, Cai L, Yu J Sterol regulatory element-binding protein cooperates with c-Myc to promote epithelial-mesenchymal transition in colorectal cancer Oncol Lett 2018;15(4):5959–65 23 Zhang Z, Shao Z, Xiong L, Yang S Inhibition of autophagy enhances cisplatin-induced apoptosis in the MG63 human osteosarcoma cell line Oncol Lett 2015;10(5):2941–6 24 Shi S, Tan P, Yan B, Gao R, Zhao J, Wang J, Guo J, Li N, Ma Z ER stress and autophagy are involved in the apoptosis induced by cisplatin in human lung cancer cells Oncol Rep 2016;35(5):2606–14 25 Chen JH, Zhang P, Chen WD, Li DD, Wu XQ, Deng R, Jiao L, Li X, Ji J, Feng GK, et al ATM-mediated PTEN phosphorylation promotes PTEN nuclear translocation and autophagy in response to DNA-damaging agents in cancer cells Autophagy 2015;11(2):239–52 26 Das CK, Mandal M, Kogel D Pro-survival autophagy and cancer cell resistance to therapy Cancer Metastasis Rev 2018;37(4):749–66 27 Hall EA, Ramsey JE, Peng Z, Hayrapetyan D, Shkepu V, O'Rourke B, Geiger W, Lam K, Verschraegen CF Novel organometallic chloroquine derivative inhibits tumor growth J Cell Biochem 2018;119(7) https://doi.org/10.1002/ jcb.26787 28 Circu M, Cardelli J, Barr M, O'Byrne K, Mills G, El-Osta H Modulating lysosomal function through lysosome membrane permeabilization or autophagy suppression restores sensitivity to cisplatin in refractory nonsmall-cell lung cancer cells PLoS One 2017;12(9):e0184922 https://doi.org/ 10.1371/journal.pone.0184922 29 Zhang Y, Liao Z, Zhang LJ, Xiao HT The utility of chloroquine in cancer therapy Curr Med Res Opin 2015;31(5):1009–13 30 Ren T, Shan J, Qing Y, Qian C, Li Q, Lu G, Li M, Li C, Peng Y, Luo H, et al Sequential treatment with AT-101 enhances cisplatin chemosensitivity in human non-small cell lung cancer cells through inhibition of apurinic/ apyrimidinic endonuclease 1-activated IL-6/STAT3 signaling pathway Drug Des Devel Ther 2014;8:2517–29 31 Hu F, Guo XL, Zhang SS, Zhao QD, Li R, Xu Q, Wei LX Suppression of p53 potentiates chemosensitivity in nutrient-deprived cholangiocarcinoma cells via inhibition of autophagy Oncol Lett 2017;14(2):1959–66 32 Kuang P, Zhou C, Li X, Ren S, Li B, Wang Y, Li J, Tang L, Zhang J, Zhao Y Proteomics-based identification of secreted protein dihydrodiol dehydrogenase as a potential biomarker for predicting cisplatin efficacy in advanced NSCLC patients Lung Cancer 2012;77(2):427–32 Page 11 of 11 33 Zhong C, Shu M, Ye J, Wang X, Liu Z, Zhao W, Zhao B, Zheng Z, Yin Z, Gao M, et al Oncogenic Ras is downregulated by ARHI and induces autophagy by Ras/AKT/mTOR pathway in glioblastoma BMC Cancer 2019;19:441 https://doi.org/10.1186/s12885-019-5643-z 34 Lock R, Roy S, Kenific CM, Su JS, Salas E, Ronen SM, Debnath J Autophagy facilitates glycolysis during Ras-mediated oncogenic transformation Mol Biol Cell 2011;22(2):165–78 35 Guo JY, Chen HY, Mathew R, Fan J, Strohecker AM, Karsli-Uzunbas G, Kamphorst JJ, Chen G, Lemons JM, Karantza V, et al Activated Ras requires autophagy to maintain oxidative metabolism and tumorigenesis Genes Dev 2011;25(5):460–70 36 Levy JM, Thompson JC, Griesinger AM, Amani V, Donson AM, Birks DK, Morgan MJ, Mirsky DM, Handler MH, Foreman NK, et al Autophagy inhibition improves chemosensitivity in BRAF(V600E) brain tumors Cancer Discov 2014;4(7):773–80 37 Karsli-Uzunbas G, Guo JY, Price S, Teng X, Laddha SV, Khor S, Kalaany NY, Jacks T, Chan CS, Rabinowitz JD, et al Autophagy is required for glucose homeostasis and lung tumor maintenance Cancer Discov 2014;4(8):914–27 38 Zhu J, Zhang C, Qing Y, Cheng Y, Jiang X, Li M, Yang Z, Wang D Genistein induces apoptosis by stabilizing intracellular p53 protein through an APE1mediated pathway Free Radical Bio Med 2015;86:209–18 Publisher’s Note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations ... resistance and that combined blockade of APE1 and autophagy enhances the therapeutic effect of cisplatin and overcomes cisplatin resistance in lung cancer In the present study, we applied quantitative... proliferation, apoptosis, and autophagy Subsequent IPA analysis revealed 72 canonical signalling pathways including the BER pathway, DNA double-strand break repair, and autophagy pathways Autophagy. .. be involved in DNA damage repair, cell proliferation, apoptosis, and autophagy Dual inhibition of APE1 and autophagy synergistically enhanced cisplatin-induced apoptosis via the regulation of APE1- p53-LC3

Ngày đăng: 06/08/2020, 05:28

Từ khóa liên quan

Mục lục

  • Abstract

    • Background

    • Methods

    • Results

    • Conclusions

    • Background

    • Methods

      • Chemicals and reagents

      • Cell line and cell culture

      • Cell viability determination

      • Quantitative proteomics

      • Quantification of cellular apoptosis

      • Quantification of cellular autophagy

      • Confocal fluorescence microscopy

      • Western blotting assay

      • RNA interference

      • Immunoprecipitation

      • Statistical analysis

      • Results

        • Overview of the proteomic response to cisplatin treatment in A549 cells

        • Cisplatin induces autophagy and apoptosis and increases APE1 expression

        • Inhibition of autophagy enhances cisplatin-induced apoptosis

        • Inhibition of APE1 stimulates autophagy in A549 cells

Tài liệu cùng người dùng

Tài liệu liên quan